Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(6): 1008-1024.e15, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35202565

RESUMEN

Vaccine-mediated immunity often relies on the generation of protective antibodies and memory B cells, which commonly stem from germinal center (GC) reactions. An in-depth comparison of the GC responses elicited by SARS-CoV-2 mRNA vaccines in healthy and immunocompromised individuals has not yet been performed due to the challenge of directly probing human lymph nodes. Herein, through a fine-needle aspiration-based approach, we profiled the immune responses to SARS-CoV-2 mRNA vaccines in lymph nodes of healthy individuals and kidney transplant recipients (KTXs). We found that, unlike healthy subjects, KTXs presented deeply blunted SARS-CoV-2-specific GC B cell responses coupled with severely hindered T follicular helper cell, SARS-CoV-2 receptor binding domain-specific memory B cell, and neutralizing antibody responses. KTXs also displayed reduced SARS-CoV-2-specific CD4 and CD8 T cell frequencies. Broadly, these data indicate impaired GC-derived immunity in immunocompromised individuals and suggest a GC origin for certain humoral and memory B cell responses following mRNA vaccination.

2.
Immunity ; 54(9): 2005-2023.e10, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525339

RESUMEN

Cell fate decisions during early B cell activation determine the outcome of responses to pathogens and vaccines. We examined the early B cell response to T-dependent antigen in mice by single-cell RNA sequencing. Early after immunization, a homogeneous population of activated precursors (APs) gave rise to a transient wave of plasmablasts (PBs), followed a day later by the emergence of germinal center B cells (GCBCs). Most APs rapidly exited the cell cycle, giving rise to non-GC-derived early memory B cells (eMBCs) that retained an AP-like transcriptional profile. Rapid decline of antigen availability controlled these events; provision of excess antigen precluded cell cycle exit and induced a new wave of PBs. Fate mapping revealed a prominent contribution of eMBCs to the MBC pool. Quiescent cells with an MBC phenotype dominated the early response to immunization in primates. A reservoir of APs/eMBCs may enable rapid readjustment of the immune response when failure to contain a threat is manifested by increased antigen availability.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Inmunidad Humoral/inmunología , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Animales , Presentación de Antígeno/inmunología , Diferenciación Celular/inmunología , Ratones , Células Plasmáticas/inmunología , Células Precursoras de Linfocitos B/inmunología
3.
Immunity ; 53(6): 1281-1295.e5, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33296685

RESUMEN

The deployment of effective vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical to eradicate the coronavirus disease 2019 (COVID-19) pandemic. Many licensed vaccines confer protection by inducing long-lived plasma cells (LLPCs) and memory B cells (MBCs), cell types canonically generated during germinal center (GC) reactions. Here, we directly compared two vaccine platforms-mRNA vaccines and a recombinant protein formulated with an MF59-like adjuvant-looking for their abilities to quantitatively and qualitatively shape SARS-CoV-2-specific primary GC responses over time. We demonstrated that a single immunization with SARS-CoV-2 mRNA, but not with the recombinant protein vaccine, elicited potent SARS-CoV-2-specific GC B and T follicular helper (Tfh) cell responses as well as LLPCs and MBCs. Importantly, GC responses strongly correlated with neutralizing antibody production. mRNA vaccines more efficiently induced key regulators of the Tfh cell program and influenced the functional properties of Tfh cells. Overall, this study identifies SARS-CoV-2 mRNA vaccines as strong candidates for promoting robust GC-derived immune responses.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Linfocitos B/inmunología , Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Centro Germinal/inmunología , SARS-CoV-2/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas Sintéticas/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Células Cultivadas , Epítopos , Humanos , Activación de Linfocitos , Polisorbatos , ARN Viral/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Escualeno , Vacunación , Vacunas de ARNm
4.
Immunity ; 53(5): 952-970.e11, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33098766

RESUMEN

Precise targeting of activation-induced cytidine deaminase (AID) to immunoglobulin (Ig) loci promotes antibody class switch recombination (CSR) and somatic hypermutation (SHM), whereas AID targeting of non-Ig loci can generate oncogenic DNA lesions. Here, we examined the contribution of G-quadruplex (G4) nucleic acid structures to AID targeting in vivo. Mice bearing a mutation in Aicda (AIDG133V) that disrupts AID-G4 binding modeled the pathology of hyper-IgM syndrome patients with an orthologous mutation, lacked CSR and SHM, and had broad defects in genome-wide AIDG133V chromatin localization. Genome-wide analyses also revealed that wild-type AID localized to MHCII genes, and AID expression correlated with decreased MHCII expression in germinal center B cells and diffuse large B cell lymphoma. Our findings indicate a crucial role for G4 binding in AID targeting and suggest that AID activity may extend beyond Ig loci to regulate the expression of genes relevant to the physiology and pathology of activated B cells.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , G-Cuádruplex , Síndrome de Inmunodeficiencia con Hiper-IgM/etiología , Síndrome de Inmunodeficiencia con Hiper-IgM/metabolismo , Mutación , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biología Computacional/métodos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Activación Enzimática , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Antígenos HLA/genética , Antígenos HLA/inmunología , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM/diagnóstico , Cambio de Clase de Inmunoglobulina/genética , Cambio de Clase de Inmunoglobulina/inmunología , Inmunofenotipificación , Activación de Linfocitos/genética , Linfoma de Células B Grandes Difuso/etiología , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Transgénicos
5.
Immunity ; 48(4): 702-715.e4, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669250

RESUMEN

Higher- or lower-affinity germinal center (GC) B cells are directed either to plasma cell or GC recycling, respectively; however, how commitment to the plasma cell fate takes place is unclear. We found that a population of light zone (LZ) GC cells, Bcl6loCD69hi expressing a transcription factor IRF4 and higher-affinity B cell receptors (BCRs) or Bcl6hiCD69hi with lower-affinity BCRs, favored the plasma cell or recycling GC cell fate, respectively. Mechanistically, CD40 acted as a dose-dependent regulator for Bcl6loCD69hi cell formation. Furthermore, we found that expression of intercellular adhesion molecule 1 (ICAM-1) and signaling lymphocytic activation molecule (SLAM) in Bcl6loCD69hi cells was higher than in Bcl6hiCD69hi cells, thereby affording more stable T follicular helper (Tfh)-GC B cell contacts. These data support a model whereby commitment to the plasma cell begins in the GC and suggest that stability of Tfh-GC B cell contacts is key for plasma cell-prone GC cell formation.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos B/citología , Antígenos CD40/metabolismo , Centro Germinal/inmunología , Lectinas Tipo C/metabolismo , Células Plasmáticas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Animales , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Molécula 1 de Adhesión Intercelular/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/biosíntesis , Linfocitos T Colaboradores-Inductores/inmunología
6.
Proc Natl Acad Sci U S A ; 121(5): e2304020121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261619

RESUMEN

Follicular regulatory T cells (Tfr) can play opposite roles in the regulation of germinal center (GC) responses. Depending on the studies, Tfr suppress or support GC and B cell affinity maturation. However, which factors determine positive vs. negative effects of Tfr on the GC B cell is unclear. In this study, we show that GC centrocytes that express MYC up-regulate expression of CCL3 chemokine that is needed for both the positive and negative regulation of GC B cells by Tfr. B cell-intrinsic expression of CCL3 contributes to Tfr-dependent positive selection of foreign Ag-specific GC B cells. At the same time, expression of CCL3 is critical for direct Tfr-mediated suppression of GC B cells that acquire cognate to Tfr nuclear proteins. Our study suggests that CCR5 and CCR1 receptors promote Tfr migration to CCL3 and highlights Ccr5 expression on the Tfr subset that expresses Il10. Based on our findings and previous studies, we suggest a model of chemotactically targeted checkpoint control of B cells undergoing positive selection in GCs by Tfr, where Tfr directly probe and license foreign antigen-specific B cells to complete their positive selection in GCs but, at the same time, suppress GC B cells that present self-antigens cognate to Tfr.


Asunto(s)
Linfocitos B , Linfocitos T Reguladores , Centro Germinal , Autoantígenos , Quimiocina CCL3
7.
Immunol Rev ; 296(1): 132-141, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32542739

RESUMEN

Influenza is one of the best examples of highly mutable viruses that are able to escape immune surveillance. Indeed, in response to influenza seasonal infection or vaccination, the majority of the induced antibodies are strain-specific. Current vaccine against the seasonal strains with the strategy of surveillance-prediction-vaccine does not cover an unmet virus strain leading to pandemic. Recently, antibodies targeting conserved epitopes on the hemagglutinin (HA) protein have been identified, albeit rarely, and they often showed broad protection. These antibody discoveries have brought the feasibility to develop a universal vaccine. Most of these antibodies bind the HA stem domain and accumulate in the memory B cell compartment. Broadly reactive stem-biased memory responses were induced by infection with antigenically divergent influenza strains and were able to eradicate these viruses, together indicating the importance of generating memory B cells expressing high-quality anti-stem antibodies. Here, we emphasize recent progress in our understanding of how such memory B cells can be generated and discuss how these advances may be relevant to the quest for a universal influenza vaccine.


Asunto(s)
Epítopos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Vacunación , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Sitios de Unión , Reacciones Cruzadas/inmunología , Epítopos/química , Epítopos/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Memoria Inmunológica , Virus de la Influenza A/clasificación , Unión Proteica , Relación Estructura-Actividad , Vacunación/métodos , Vacunología
8.
Proc Natl Acad Sci U S A ; 117(22): 12295-12305, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32424104

RESUMEN

The mechanisms that regulate germinal center (GC) B cell responses in the spleen are not fully understood. Here we use a combination of pharmacologic and genetic approaches to delete SIGN-R1+ marginal zone (MZ) macrophages and reveal their specific contribution to the regulation of humoral immunity in the spleen. We find that while SIGN-R1+ macrophages were not essential for initial activation of B cells, they were required for maturation of the response and development of GC B cells. These defects could be corrected when follicular helper T (Tfh) cells were induced before macrophage ablation or when Tfh responses were enhanced. Moreover, we show that in the absence of SIGN-R1+ macrophages, DCIR2+ dendritic cells (DCs), which play a key role in priming Tfh responses, were unable to cluster to the interfollicular regions of the spleen and were instead displaced to the MZ. Restoring SIGN-R1+ macrophages to the spleen corrected positioning of DCIR2+ DCs and rescued the GC B cell response. Our study reveals a previously unappreciated role for SIGN-R1+ macrophages in regulation of the GC reaction and highlights the functional specification of macrophage subsets in the MZ compartment.


Asunto(s)
Linfocitos B/inmunología , Moléculas de Adhesión Celular/inmunología , Centro Germinal/inmunología , Lectinas Tipo C/inmunología , Macrófagos/inmunología , Receptores de Superficie Celular/inmunología , Bazo/inmunología , Animales , Moléculas de Adhesión Celular/genética , Lectinas Tipo C/genética , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Superficie Celular/genética , Linfocitos T Colaboradores-Inductores
9.
Immunol Rev ; 292(1): 120-138, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31631359

RESUMEN

The current concepts for development of autoreactive B cells in SLE (systemic lupus erythematosus) focus on extrinsic stimuli and factors that provoke B cells into tolerance loss. Traditionally, major tolerance loss pathways are thought to be regulated by factors outside the B cell including autoantigen engagement of the B-cell receptor (BCR) with simultaneous type I interferon (IFN) produced by dendritic cells, especially plasmacytoid dendritic cells (pDCs). Later, in autoreactive follicles, B-cells encounter T-follicular helper cells (Tfh) that produce interleukin (IL)-21, IL-4 and pathogenic cytokines, IL-17 and IFN gamma (IFNÉ£). This review discusses these mechanisms and also highlights recent advances pointing to the peripheral transitional B-cell stage as a major juncture where transient autocrine IFNß expression by developing B-cells imprints a heightened susceptibility to external factors favoring differentiation into autoantibody-producing plasmablasts. Recent studies highlight transitional B-cell heterogeneity as a determinant of intrinsic resistance or susceptibility to tolerance loss through the shaping of B-cell responsiveness to cytokines and other environment factors.


Asunto(s)
Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Tolerancia Inmunológica/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Lupus Eritematoso Sistémico/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología
10.
Eur J Immunol ; 51(11): 2555-2567, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34324199

RESUMEN

Upon antigen exposure, activated B cells in antigen-draining lymphoid organs form microanatomical structures, called germinal centers (GCs), where affinity maturation occurs. Within the GC microenvironment, GC B cells undergo proliferation and B cell receptor (BCR) genes somatic hypermutation in the dark zone (DZ), and affinity-based selection in the light zone (LZ). In the current paradigm of GC dynamics, high-affinity LZ B cells may be selected by cognate T- follicular helper cells to either differentiate into plasma cells or memory B cells, or re-enter the DZ and initiate a new round of proliferation and BCR diversification, before migrating back to the LZ. Given the diversity of cell states and potential cell fates that GC B cells may adopt, the two-state DZ-LZ paradigm has been challenged by studies that explored GC B-cell heterogeneity with a variety of single-cell technologies. Here, we review studies and single-cell technologies which have allowed to refine the working model of GC B-cell cellular and molecular heterogeneity during affinity maturation. This review also covers the use of single-cell quantitative data for mathematical modeling of GC reactions, and the application of single-cell genomics to the study of GC-derived malignancies.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Activación de Linfocitos/inmunología , Animales , Humanos , Modelos Teóricos , Análisis de la Célula Individual
11.
Eur J Immunol ; 50(7): 1067-1077, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32133634

RESUMEN

Stearoyl-CoA desaturases (SCD) are endoplasmic reticulum (ER)-associated enzymes that catalyze the synthesis of the monounsaturated fatty acids (MUFAs). As such, SCD play important roles in maintaining the intracellular balance between saturated fatty acid (SFAs) and MUFAs. The roles of SCD in CD4+ T-helper cell responses are currently unexplored. Here, we have found that murine and human follicular helper T (TFH ) cells express higher levels of SCD compared to non-TFH cells. Further, the expression of SCD in TFH cells is dependent on the TFH lineage-specification transcription factor BCL6. We found that the inhibition of SCD impaired TFH cell maintenance and shifted the balance between TFH and follicular regulatory T (TFR ) cells in the spleen. Consequently, SCD inhibition dampened germinal center B-cell responses following influenza immunization. Mechanistically, we found that SCD inhibition led to increased ER stress and enhanced TFH cell apoptosis in vitro and in vivo. These results reveal a possible link between fatty acid metabolism and cellular and humoral responses induced by immunization or potentially, autoimmunity.


Asunto(s)
Apoptosis/inmunología , Linfocitos B/inmunología , Centro Germinal/inmunología , Bazo/inmunología , Estearoil-CoA Desaturasa/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/citología , Centro Germinal/citología , Humanos , Ratones , Ratones Noqueados , Bazo/citología , Estearoil-CoA Desaturasa/genética , Linfocitos T Reguladores/citología
12.
Int Arch Allergy Immunol ; 182(3): 195-209, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33486489

RESUMEN

We report perhaps the most comprehensive study of subsets of CD4+ and CD8+ and subsets of B cells in a mild symptomatic SARS-CoV-2+ immunocompetent patient and a common variable immunodeficiency disease (CVID) patient who had normal absolute lymphocyte counts and remained negative for SARS-CoV-2 IgG antibodies. Naïve (TN), central memory (TCM), effector memory (TEM), and terminally differentiated effector memory (TEMRA) subsets of CD4+ and CD8+ T cells, subsets of T follicular helper cells (cTFH, TFH1, TFH2, TFH17, TFH1/TFH17, and TFR), CD4 Treg, CD8 Treg, mature B cells, transitional B cells, marginal zone B cells, germinal center (GC) B cells, CD21low B cells, antibody-secreting cells (plasmablasts), and Breg cells were examined in patients and age-matched controls with appropriate monoclonal antibodies and isotype controls using multicolor flow cytometry. Different patterns of abnormalities (often contrasting) were observed in the subsets of CD4+ T, CD8+ T, B-cell subsets, and regulatory lymphocytes among the immunocompetent patient and CVID patient as compared to corresponding healthy controls. Furthermore, when data were analyzed between the 2 patients, the immunocompetent patient demonstrated greater changes in various subsets as compared to the CVID patient. These data demonstrate different immunological responses to SARS-CoV-2 infection in an immunocompetent patient and the CVID patient. A marked decrease in GC B cells and plasmablasts may be responsible for failure to make SARS-CoV-2 antibodies. The lack of SARS-CoV-2 antibodies with mild clinical disease suggests an important role of T-cell response in defense against SARS-CoV-2 infection.


Asunto(s)
COVID-19/inmunología , Inmunodeficiencia Variable Común/inmunología , SARS-CoV-2/inmunología , Subgrupos de Linfocitos T/inmunología , Adulto , Subgrupos de Linfocitos B/inmunología , Femenino , Humanos , Inmunocompetencia , Masculino , Persona de Mediana Edad
13.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626686

RESUMEN

Live viral vaccines elicit protective, long-lived humoral immunity, but the underlying mechanisms through which this occurs are not fully elucidated. Generation of affinity matured, long-lived protective antibody responses involve close interactions between T follicular helper (TFH) cells, germinal center (GC) B cells, and T follicular regulatory (TFR) cells. We postulated that escalating concentrations of antigens from replicating viruses or live vaccines, spread through the hematogenous route, are essential for the induction and maintenance of long-lived protective antibody responses. Using replicating and poorly replicating or nonreplicating orthopox and influenza A viruses, we show that the magnitude of TFH cell, GC B cell, and neutralizing antibody responses is directly related to virus replicative capacity. Further, we have identified that both lymphoid and circulating TFH:TFR cell ratios during the peak GC response can be used as an early predictor of protective, long-lived antibody response induction. Finally, administration of poorly or nonreplicating viruses to allow hematogenous spread generates significantly stronger TFH:TFR ratios and robust TFH, GC B cell and neutralizing antibody responses.IMPORTANCE Neutralizing antibody response is the best-known correlate of long-term protective immunity for most of the currently licensed clinically effective viral vaccines. However, the host immune and viral factors that are critical for the induction of robust and durable antiviral humoral immune responses are not well understood. Our study provides insight into the dynamics of key cellular mediators of germinal center reaction during live virus infections and the influence of viral replicative capacity on the magnitude of antiviral antibody response and effector function. The significance of our study lies in two key findings. First, the systemic spread of even poorly replicating or nonreplicating viruses to mimic the spread of antigens from replicating viruses due to escalating antigen concentration is fundamental to the induction of durable antibody responses. Second, the TFH:TFR ratio may be used as an early predictor of protective antiviral humoral immune responses long before memory responses are generated.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Antígenos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Replicación Viral/inmunología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Línea Celular , Chlorocebus aethiops , Perros , Centro Germinal/inmunología , Inmunidad Humoral/inmunología , Células de Riñón Canino Madin Darby , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Vacunas Atenuadas/inmunología
14.
Immunol Rev ; 269(1): 194-211, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26683154

RESUMEN

Autoimmune diseases are characterized by adaptive immune responses against self-antigens, including humoral responses resulting in the production of autoantibodies. Autoantibodies generate inflammation by activating complement and engaging Fcγ receptors (FcγRs). The inhibitory receptor FcγRIIB plays a central role in regulating the generation of autoantibodies and their effector functions, which include activation of innate immune cells and the cellular arm of the adaptive immune system, via effects on antigen presentation to CD4 T cells. Polymorphisms in FcγRIIB have been associated with susceptibility to autoimmunity but protection against infections in humans and mice. In the last few years, new mechanisms by which FcγRIIB controls the adaptive immune response have been described. Notably, FcγRIIB has been shown to regulate germinal center B cells and dendritic cell migration, with potential impact on the development of autoimmune diseases. Recent work has also highlighted the implication of FcγRIIB on the regulation of the innate immune system, via inhibition of Toll-like receptor- and complement receptor-mediated activation. This review will provide an update on the role of FcγRIIB in adaptive immune responses in autoimmunity, and then focus on their emerging function in innate immunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Receptores de IgG/inmunología , Inmunidad Adaptativa , Animales , Autoinmunidad , Predisposición Genética a la Enfermedad , Homeostasis , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Inmunomodulación , Ratones , Polimorfismo Genético , Receptores de IgG/genética
15.
Cytometry A ; 95(4): 442-449, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30838773

RESUMEN

CD40 expression is required for germinal center (GC) formation and function, but the kinetics and magnitude of signaling following CD40 engagement remain poorly characterized in human B cells undergoing GC reactions. Here, differences in CD40 expression and signaling responses were compared across differentiation stages of mature human tonsillar B cells. A combination of mass cytometry and phospho-specific flow cytometry was used to quantify protein expression and CD40L-induced signaling in primary human naïve, GC, and memory B cells. Protein expression signatures of cell subsets were quantified using viSNE and Marker Enrichment Modeling (MEM). This approach revealed enriched expression of CD40 protein in GC B cells, compared to naïve and memory B cells. Despite this, GC B cells responded to CD40L engagement with lower phosphorylation of NFκB p65 during the first 30 min following CD40L activation. Before CD40L stimulation, GC B cells expressed higher levels of suppressor protein IκBα than naïve and memory B cells. Following CD40 activation, IκBα was rapidly degraded and reached equivalently low levels in naïve, GC, and memory B cells at 30 min following CD40L. Quantifying CD40 signaling responses as a function of bound ligand revealed a correlation between bound CD40L and degree of induced NFκB p65 phosphorylation, whereas comparable IκBα degradation occurred at all measured levels of CD40L binding. These results characterize cell-intrinsic signaling differences that exist in mature human B cells undergoing GC reactions. © 2019 International Society for Advancement of Cytometry.


Asunto(s)
Linfocitos B/fisiología , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Centro Germinal/citología , Memoria Inmunológica , Linfocitos B/citología , Linfocitos B/metabolismo , Ligando de CD40/fisiología , Células Cultivadas , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , FN-kappa B/metabolismo , Fosforilación , Transducción de Señal/inmunología
16.
Eur J Immunol ; 47(9): 1443-1456, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28643847

RESUMEN

NKp46/NCR1 is an activating NK-cell receptor implicated in the control of various viral and bacterial infections. Recent findings also suggest that it plays a role in shaping the adaptive immune response to pathogens. Using NCR1-deficient (NCR1gfp/gfp ) mice, we provide evidence for the role of NCR1 in antibody response to mouse cytomegalovirus infection (MCMV). The absence of NCR1 resulted in impaired maturation, function and NK-cell migration to regional lymph nodes. In addition, CD4+ T-cell activation and follicular helper T-cell (Tfh) generation were reduced, leading to inferior germinal center (GC) B-cell maturation. As a consequence, NCR1gfp/gfp mice produced lower amounts of MCMV-specific antibodies upon infection, which correlated with lower number of virus-specific antibody secreting cells in analyzed lymph nodes.


Asunto(s)
Antígenos Ly/metabolismo , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Centro Germinal/inmunología , Infecciones por Herpesviridae/inmunología , Células Asesinas Naturales/inmunología , Muromegalovirus/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Animales , Anticuerpos Antivirales/sangre , Antígenos Ly/genética , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Inmunidad Humoral , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/genética
17.
J Virol ; 91(3)2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27852854

RESUMEN

Rabies remains a public health threat in most parts of the world, and approximately 99% of the cases are transmitted by dogs. There is an urgent need to develop an efficacious and affordable vaccine to control canine-transmitted rabies in developing countries. Our previous studies demonstrate that overexpression of chemokines/cytokines such as CCL-3 (MIP-1α) and granulocyte-macrophage colony-stimulating factor (GM-CSF) can enhance the immunogenicity of rabies vaccines. In the present study, the chemokine CXCL13 was inserted into the genome of the recombinant rabies virus (rRABV) strain LBNSE, and the effect of the chemokine CXCL13 on the immunogenicity of RABV was investigated. It was found that LBNSE-CXCL13 recruited follicular helper T (Tfh) and germinal center (GC) B cells, promoted the formation of GCs, and increased the population of plasma cells in immunized mice. Further studies showed that mice immunized with LBNSE-CXCL13 produced more rabies virus-neutralizing antibodies (VNAs) and developed better protection than those immunized with the parent virus LBNSE or the GM-CSF-expressing RABV (LBNSE-GM-CSF). Collectively, these findings provide a better understanding of the role of CXCL13 expression in the immunogenicity of the RABV, which may help in designing more-efficacious rabies vaccines. IMPORTANCE: Rabies is endemic in most parts of the world, and more effort is needed to develop affordable and effective vaccines to control or eliminate this disease. The chemokine CXCL13 recruits both Tfh and B cells, which is essential for the homing of Tfh cells and the development of B cell follicles. In this study, the effect of the overexpression of CXCL13 on the immunogenicity of the RABV was evaluated in a mouse model. We found that CXCL13 expression promoted humoral immunity by recruiting Tfh and GC B cells, facilitating the formation of GCs, and increasing the number of plasma cells. As expected, the overexpression of CXCL13 resulted in enhanced virus-neutralizing antibody (VNA) production and protection against a virulent RABV challenge. These findings provide a better understanding of the role of CXCL13 in RABV-induced immune responses, which will help in designing more efficacious rabies vaccines.


Asunto(s)
Linfocitos B/inmunología , Quimiocina CXCL13/genética , Expresión Génica , Centro Germinal/inmunología , Inmunidad Humoral , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/metabolismo , Quimiocina CXCL13/sangre , Quimiotaxis/inmunología , Cricetinae , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Rabia/inmunología , Rabia/prevención & control , Virus de la Rabia/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
18.
Proc Natl Acad Sci U S A ; 112(48): E6644-53, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26627255

RESUMEN

Switched antibody classes are important for efficient immune responses. Aberrant antibody production to otherwise harmless antigens may result in autoimmunity. The protein kinase fms-like tyrosine kinase 3 receptor (Flt3) has an important role during early B-cell development, but the role of Flt3 in peripheral B cells has not been assessed before. Herein we describe a previously unappreciated role for Flt3 in IgG1 class-switch recombination (CSR) and production. We show that Flt3 is reexpressed on B-cell lymphoma 6(+) germinal center B cells in vivo and following LPS activation of peripheral B cells in vitro. Absence of Flt3 signaling in Flt3 ligand-deficient mice results in impaired IgG1 CSR and accumulation of IgM-secreting plasma cells. On activated B cells, Flt3 is coexpressed and functions in synergy with the common-gamma chain receptor family. B cells from Flt3 ligand-deficient mice have impaired IL-4R signaling, with reduced phosphorylation of signal transducer and activator of transcription (Stat) 6, and demonstrate a failure to initiate CSR to IgG1 with low expression of γ1 germ-line transcripts, resulting in impaired IgG1 production. Thus, functional synergy between Flt3 and IL-4R signaling is critical for Stat-mediated regulation of sterile γ1 germ-line transcripts and CSR to IgG1.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/inmunología , Tirosina Quinasa 3 Similar a fms/fisiología , Animales , Apoptosis , Regulación de la Expresión Génica , Inmunoglobulina M/inmunología , Ligandos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Plasmáticas/inmunología , Receptores de Interleucina-4/metabolismo , Transducción de Señal , Tirosina Quinasa 3 Similar a fms/genética
19.
Cell Immunol ; 294(1): 1-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25656449

RESUMEN

Germinal center (GC) reaction is a T cell-dependent process in which activated B cells undergo clonal expansion and functional maturation to produce high affinity antibodies and differentiate into memory B cells(1). Here we demonstrate a new role of bromodomain and extraterminal domain (BET) protein BRD4 in GC B cell development. We found that during B cell differentiation stage there was an elevated expression of BRD4 in GC B cells and inhibition of BRD4 by small molecule inhibitors led to the suppression of GC formation and correspondent antibody responses in a Td antigen immunization model. At the molecular level, we found that the effects of BRD4 in primary GC B cell differentiation and B cell lymphoma were mediated through the impaired phosphorylation and translocation of NF-κBp65 and further down-regulation of B-cell lymphoma 6 (Bcl6) expression. Thus this study reveals a novel function of BRD4 in controlling the GC B cell development pathway.


Asunto(s)
Linfocitos B/citología , Centro Germinal/inmunología , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Factor de Transcripción ReIA/inmunología , Factores de Transcripción/fisiología , Animales , Anticuerpos/sangre , Anticuerpos/inmunología , Azepinas/farmacología , Linfocitos B/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Proteínas de Ciclo Celular , Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Factores Reguladores del Interferón/biosíntesis , Activación de Linfocitos/inmunología , Linfoma de Células B/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Nucleoproteínas/inmunología , Factor de Transcripción PAX5/biosíntesis , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-6/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-6/genética , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Transducción de Señal/inmunología , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/genética , Triazoles/farmacología
20.
Pediatr Hematol Oncol ; 32(4): 229-38, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25252151

RESUMEN

Pediatric Hodgkin lymphoma (HL) comprises approximately a fifth of all patients with HL in India. Seventy-four cases of pediatric classical Hodgkin Lymphoma (cHL) from a regional cancer center in southern India were analyzed on a tissue microarray (TMA) for the stage of B-cell differentiation of the Hodgkin/Reed Sternberg (HRS) cell by immunohistochemistry (IHC) using CD10, bcl6, MUM1/IRF4, and CD 138. Fifty-two of seventy-four (70.3%) cases were of late germinal center/early post-germinal center phenotype (CD10-/bcl6-/MUM1+/CD138-). Epstein-Barr virus (EBV) association using Epstein-Barr virus encoded RNA (EBER) RISH and EBV-LMP1 immunohistochemistry (IHC) revealed an EBV association of 93%. Tumor-associated macrophages (TAM) in the microenvironment were also assessed on the TMA by CD68 IHC, and most cases (59.7%) showed >25% TAMs, with no case showing ≤5%. These findings indicate that pediatric cHL in India is a tumor, predominantly, of late germinal center/early post-germinal center B cells, is almost invariably EBV associated, and with a high number of TAMs in the microenvironment. This latter finding suggests that criteria other than TAM scores need to be developed for risk stratification of pediatric EBV-associated HL especially in developing countries.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Enfermedad de Hodgkin/metabolismo , Macrófagos/metabolismo , Células de Reed-Sternberg/metabolismo , Adolescente , Niño , Preescolar , Infecciones por Virus de Epstein-Barr/epidemiología , Infecciones por Virus de Epstein-Barr/patología , Femenino , Enfermedad de Hodgkin/epidemiología , Enfermedad de Hodgkin/patología , Humanos , India , Macrófagos/patología , Masculino , Células de Reed-Sternberg/patología , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA